Skip to main content
Log in

RAGE deficiency ameliorates autoimmune hepatitis involving inhibition of IL-6 production via suppressing protein Arid5a in mice

  • Research
  • Published:
Clinical and Experimental Medicine Aims and scope Submit manuscript

Abstract

Activation of T cells and pro-inflammatory cytokines are essential for human autoimmune hepatitis. RAGE is one of the receptors for the inflammatory alarm molecule high mobility group box 1 (HMGB1), and it is involved in autoimmune hepatitis. However, the molecular mechanism of RAGE in the context of autoimmune hepatitis remains elusive. This study aimed to identify the function and mechanism of RAGE in autoimmune hepatitis. The role and underlying mechanisms of RAGE signaling-driven immune inflammatory response in ConA-induced experimental hepatitis were examined using the RAGE-deficient mice. We found that the RAGE deficiency protected the mouse from liver inflammatory injury caused by the ConA challenge. mRNA expression of VCAM-1, IL-6, and TNF-α within the livers is markedly decreased in RAGE-deficient mice compared to wild-type mice. In parallel, RAGE deficiency leads to reduced levels of the serum pro-inflammatory cytokines IL-6 and TNF-α as compared with wild-type control mice. RAGE-deficient mice exhibit increased hepatic NK cells and decreased CD4+ T cells compared with wild-type control mice. Notably, in vivo blockade of IL-6 in wild-type mice significantly protected mice from ConA-induced hepatic injury. Furthermore, RAGE deficiency impaired IL-6 production and was associated with decreased expression of Arid5a in liver tissues, a half-life IL-6 mRNA regulator. RAGE signaling is important in regulating the development of autoimmune hepatitis. Immune regulation of RAGE may represent a novel therapeutic strategy to prevent immune-mediated liver injury.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Availability of data and materials

All data used during the current study available from the corresponding author on reasonable request.

Abbreviations

RAGE:

The receptor for advanced glycation end-product

AGEs:

Advanced glycation end products

HMGB1:

High mobility group box 1

AIH:

Autoimmune hepatitis patients

EN-RAGE:

Extracellular newly identified receptor for advanced glycation end products binding protein

sRAGE:

Soluble RAGE

ConA:

Concanavalin A

I/R:

Hepatic ischemia/reperfusion

ALT:

Alanine aminotransferase

AST:

Aspartate aminotransferase

MPO:

Myeloperoxidase

ICAM-1:

Intercellular adhesion molecule-1

VCAM-1:

Vascular cell adhesion molecule-1

WT:

Wild-type mice

Arid5a:

AT-rich interactive domain-containing protein 5a

References

  1. Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol. 2010;28:367–88.

    Article  CAS  PubMed  Google Scholar 

  2. Yatime L, Andersen GR. The specificity of DNA recognition by the RAGE receptor. J Exp Med. 2014;211(5):749–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Xie J, Méndez JD, Méndez-Valenzuela V, Aguilar-Hernández MM. Cellular signalling of the receptor for advanced glycation end products (RAGE). Cell Signal. 2013;25(11):2185–97.

    Article  CAS  PubMed  Google Scholar 

  4. Jangde N, Ray R, Rai V. RAGE and its ligands: from pathogenesis to therapeutics. Crit Rev Biochem Mol Biol. 2020;55(6):555–75.

    Article  CAS  PubMed  Google Scholar 

  5. Gong Q, Zhang H, Li JH, et al. High-mobility group box 1 exacerbates concanavalin A-induced hepatic injury in mice. J Mol Med. 2010;88(12):1289–98.

    Article  CAS  PubMed  Google Scholar 

  6. Hua S, Ma M, Fei X, Zhang Y, Gong F, Fang M. Glycyrrhizin attenuates hepatic ischemia-reperfusion injury by suppressing HMGB1-dependent GSDMD-mediated kupffer cells pyroptosis. Int Immunopharmacol. 2019;68:145–55.

    Article  CAS  PubMed  Google Scholar 

  7. Ge X, Arriazu E, Magdaleno F, et al. High mobility group box-1 drives fibrosis progression signaling via the receptor for advanced glycation end products in mice. Hepatology. 2018;68(6):2380–404.

    Article  CAS  PubMed  Google Scholar 

  8. Jhun J, Lee S, Kim H, et al. HMGB1/RAGE induces IL-17 expression to exaggerate inflammation in peripheral blood cells of hepatitis B patients. J Transl Med. 2015;13:310.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Wu R, Liu Y, Yan R, Liu X, Duan L. Assessment of EN-RAGE, sRAGE and EN-RAGE/sRAGE as potential biomarkers in patients with autoimmune hepatitis. J Transl Med. 2020;18(1):384.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yang L, Zhou L, Wang X, Wang W, Wang J. Inhibition of HMGB1 involved in the protective of salidroside on liver injury in diabetes mice. Int Immunopharmacol. 2020;89(Pt A):106987.

    Article  CAS  PubMed  Google Scholar 

  11. Zeng S, Feirt N, Goldstein M, et al. Blockade of receptor for advanced glycation end product (RAGE) attenuates ischemia and reperfusion injury to the liver in mice. Hepatology. 2004;39(2):422–32.

    Article  CAS  PubMed  Google Scholar 

  12. Moy KA, Jiao L, Freedman ND, et al. Soluble receptor for advanced glycation end products and risk of liver cancer. Hepatology. 2013;57(6):2338–45.

    Article  CAS  PubMed  Google Scholar 

  13. Nyati KK, Masuda K, Zaman MM, et al. TLR4-induced NF-κB and MAPK signaling regulate the IL-6 mRNA stabilizing protein Arid5a. Nucleic Acids Res. 2017;45(5):2687–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Khambu B, Yan S, Huda N, Yin XM. Role of High-mobility group box-1 in liver pathogenesis. Int J Mol Sci. 2019;20(21):5314.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Andersson U, Tracey KJ. HMGB1 is a therapeutic target for sterile inflammation and infection. Annu Rev Immunol. 2011;29:139–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zhong H, Li X, Zhou S, et al. Interplay between RAGE and TLR4 Regulates HMGB1-induced inflammation by promoting cell surface expression of RAGE and TLR4. J Immunol. 2020;205(3):767–75.

    Article  CAS  PubMed  Google Scholar 

  17. Chen J, Duan L, Xiong A, et al. Blockade of IL-33 ameliorates Con A-induced hepatic injury by reducing NKT cell activation and IFN-γ production in mice. J Mol Med. 2012;90(12):1505–15.

    Article  CAS  PubMed  Google Scholar 

  18. Park EJ, Lee JH, Yu GY, et al. Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell. 2010;140(2):197–208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Xiang WQ, Feng WF, Ke W, Sun Z, Chen Z, Liu W. Hepatitis B virus X protein stimulates IL-6 expression in hepatocytes via a MyD88-dependent pathway. J Hepatol. 2011;54(1):26–33.

    Article  CAS  PubMed  Google Scholar 

  20. Hammerich L, Heymann F, Tacke F. Role of IL-17 and Th17 cells in liver diseases. Clin Dev Immunol. 2011;2011:345803.

    Article  PubMed  Google Scholar 

  21. Masuda K, Ripley B, Nishimura R, et al. Arid5a controls IL-6 mRNA stability, which contributes to elevation of IL-6 level in vivo. Proc Natl Acad Sci USA. 2013;110(23):9409–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Masuda K, Ripley B, Nyati KK, et al. Arid5a regulates naive CD4+ T cell fate through selective stabilization of Stat3 mRNA. J Exp Med. 2016;213(4):605–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Nyati KK, Agarwal RG, Sharma P, Kishimoto T. Arid5a regulation and the roles of Arid5a in the inflammatory response and disease. Front Immunol. 2019;10:2790.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Weinhage T, Wirth T, Schütz P, et al. The receptor for advanced glycation endproducts (RAGE) contributes to severe inflammatory liver lnjury in mice. Front Immunol. 2020;11:1157.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Mr. Yong Xu and Zhihui Liang for technical assistance with flow cytometry.

Funding

This work was supported by grants from the National Natural Science Foundation of China (91542110, 81373167 to M. Fang).

Author information

Authors and Affiliations

Authors

Contributions

XL, DF and MF wrote the main manuscript text. XL, SH, DF, XF, ZT, FZ, WW, MF prepared Figs. 1, 2, 3, 4, 5, 6, 7, 8. All authors reviewed the manuscript.

Corresponding author

Correspondence to Min Fang.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Consent for publication

Not applicable.

Ethics approval and consent to participate

All animal protocols were approved by the Animal Research Committee at Tongji Medical College Animal Care and Use Committee (Animal permit number S2146).

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, X., Hua, S., Fang, D. et al. RAGE deficiency ameliorates autoimmune hepatitis involving inhibition of IL-6 production via suppressing protein Arid5a in mice. Clin Exp Med 23, 2167–2179 (2023). https://doi.org/10.1007/s10238-022-00960-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10238-022-00960-8

Keywords

Navigation